Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 9(1): 4870, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30451821

RESUMO

RORγt controls the differentiation of TH17 cells, which are mediators of autoimmune conditions such as experimental autoimmune encephalomyelitis (EAE). RORγt also regulates thymocyte development and lymph node genesis. Here we show that the function of RORγt is regulated by its sumoylation. Loss of Sumo3, but not Sumo1, dampens TH17 differentiation and delays the progression of thymic CD8+ immature single-positive cells (ISPs). RORγt is SUMO3-modified by E3 ligase PIAS4 at lysine 31 (K31), and the mutation of K31 to arginine in mice prevents RORγt sumoylation, leading to impaired TH17 differentiation, resistance to TH17-mediated EAE, accumulation of thymic ISPs, and a lack of Peyer's patches. Mechanistically, sumoylation of RORγt-K31 recruits histone acetyltransferase KAT2A, which stabilizes the binding of SRC1 to enhance RORγt transcription factor activity. This study thus demonstrates that sumoylation is a critical mechanism for regulating RORγt function, and reveals new drug targets for preventing TH17-mediated autoimmunity.


Assuntos
Encefalomielite Autoimune Experimental/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Processamento de Proteína Pós-Traducional , Células Th17/imunologia , Timócitos/microbiologia , Timo/imunologia , Ubiquitinas/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Hematopoese/genética , Hematopoese/imunologia , Linfonodos/imunologia , Linfonodos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Coativador 1 de Receptor Nuclear/genética , Coativador 1 de Receptor Nuclear/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/patologia , Proteína SUMO-1/deficiência , Proteína SUMO-1/genética , Proteína SUMO-1/imunologia , Sumoilação , Células Th17/patologia , Timócitos/imunologia , Timócitos/patologia , Timo/patologia , Ubiquitinas/deficiência , Ubiquitinas/imunologia , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/imunologia
2.
Lab Invest ; 98(6): 799-813, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29472640

RESUMO

The intestinal epithelium constitutes a crucial defense to the potentially life-threatening effects of gut microbiota. However, due to a complex underlying vasculature, hypoperfusion and resultant tissue ischemia pose a particular risk to function and integrity of the epithelium. The small ubiquitin-like modifier (SUMO) conjugation pathway critically regulates adaptive responses to metabolic stress and is of particular significance in the gut, as inducible knockout of the SUMO-conjugating enzyme Ubc9 results in rapid intestinal epithelial disintegration. Here we analyzed the pattern of individual SUMO isoforms in intestinal epithelium and investigated their roles in intestinal ischemia/reperfusion (I/R) damage. Immunostaining revealed that epithelial SUMO2/3 expression was almost exclusively limited to crypt epithelial nuclei in unchallenged mice. However, intestinal I/R or overexpression of Ubc9 caused a remarkable enhancement of epithelial SUMO2/3 staining along the crypt-villus axis. Unexpectedly, a similar pattern was found in SUMO1 knockout mice. Ubc9 transgenic mice, but also SUMO1 knockout mice were protected from I/R injury as evidenced by better preserved barrier function and blunted inflammatory responses. PCR array analysis of microdissected villus-tip epithelia revealed a specific epithelial contribution to reduced inflammatory responses in Ubc9 transgenic mice, as key chemotactic signaling molecules such as IL17A were significantly downregulated. Together, our data indicate a critical role particularly of the SUMO2/3 isoforms in modulating responses to I/R and provide the first evidence that SUMO1 deletion activates a compensatory process that protects from ischemic damage.


Assuntos
Mucosa Intestinal/irrigação sanguínea , Traumatismo por Reperfusão/prevenção & controle , Proteína SUMO-1/fisiologia , Enzimas de Conjugação de Ubiquitina/fisiologia , Animais , Quimiocinas/análise , Mucosa Intestinal/química , Microdissecção e Captura a Laser , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína SUMO-1/deficiência , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/análise , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Enzimas de Conjugação de Ubiquitina/genética , Ubiquitinas/análise , Ubiquitinas/fisiologia
3.
PLoS One ; 11(2): e0150324, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26927328

RESUMO

In previous work, small ubiquitin-like modifier (SUMO) in hemocytes of Chinese shrimp Fenneropenaeus chinensis was found to be up-regulated post-white spot syndrome virus (WSSV) infection using proteomic approach. However, the role of SUMO in viral infection is still unclear. In the present work, full length cDNAs of SUMO (FcSUMO) and SUMO-conjugating enzyme E2 UBC9 (FcUBC9) were cloned from F. chinensis using rapid amplification of cDNA ends approach. The open reading frame (ORF) of FcSUMO encoded a 93 amino acids peptide with the predicted molecular weight (M.W) of 10.55 kDa, and the UBC9 ORF encoded a 160 amino acids peptide with the predicted M.W of 18.35 kDa. By quantitative real-time RT-PCR, higher mRNA transcription levels of FcSUMO and FcUBC9 were detected in hemocytes and ovary of F. chinensis, and the two genes were significantly up-regulated post WSSV infection. Subsequently, the recombinant proteins of FcSUMO and FcUBC9 were expressed in Escherichia coli BL21 (DE3), and employed as immunogens for the production of polyclonal antibody (PAb). Indirect immunofluorescence assay revealed that the FcSUMO and UBC9 proteins were mainly located in the hemocytes nuclei. By western blotting, a 13.5 kDa protein and a 18.7 kDa protein in hemocytes were recognized by the PAb against SUMO or UBC9 respectively. Furthermore, gene silencing of FcSUMO and FcUBC9 were performed using RNA interference, and the results showed that the number of WSSV copies and the viral gene expressions were inhibited by knockdown of either SUMO or UBC9, and the mortalities of shrimp were also reduced. These results indicated that FcSUMO and FcUBC9 played important roles in WSSV infection.


Assuntos
Penaeidae/virologia , Proteína SUMO-1/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Vírus da Síndrome da Mancha Branca 1/fisiologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar/genética , Regulação Enzimológica da Expressão Gênica , Inativação Gênica , Hemócitos/metabolismo , Dados de Sequência Molecular , Penaeidae/enzimologia , Penaeidae/genética , Filogenia , Interferência de RNA , Proteína SUMO-1/química , Proteína SUMO-1/deficiência , Proteína SUMO-1/genética , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/genética
4.
Arch Virol ; 161(1): 141-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26449956

RESUMO

Infection by hepatitis C virus (HCV) is a major public-health problem. Chronic infection often leads to cirrhosis, steatosis, and hepatocellular carcinoma. The life cycle of HCV depends on the host cell machinery and involves intimate interaction between viral and host proteins. However, the role of host proteins in the life cycle of HCV remains poorly understood. Here, we identify the small ubiquitin-related modifier (SUMO1) as a key host factor required for HCV replication. We performed a series of cell biology and biochemistry experiments using the HCV JFH-1 (Japanese fulminate hepatitis 1) genotype 2a strain, which produces infectious particles and recapitulates all the steps of the HCV life cycle. We observed that SUMO1 is upregulated in Huh7.5 infected cells. Reciprocally, SUMO1 was found to regulate the expression of viral core protein. Moreover, knockdown of SUMO1 using specific siRNA influenced the accumulation of lipid droplets and reduced HCV replication as measured by qRT-PCR. Thus, we identify SUMO1 as a key host factor required for HCV replication. To our knowledge, this is the first report showing that SUMO1 regulates lipid droplets in the context of viral infection. Our report provides a meaningful insight into how HCV replicates and interacts with host proteins and is of significant importance for the field of HCV and RNA viruses.


Assuntos
Hepacivirus/fisiologia , Hepatite C/metabolismo , Gotículas Lipídicas/metabolismo , Proteína SUMO-1/deficiência , Replicação Viral , Linhagem Celular , Genótipo , Hepacivirus/genética , Hepatite C/genética , Hepatite C/virologia , Interações Hospedeiro-Patógeno , Humanos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína SUMO-1/genética , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo
5.
Sci Rep ; 5: 9011, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25757417

RESUMO

The small ubiquitin-related modifier (SUMO) participates in various cellular processes, including maintenance of genome integrity, nuclear transport, transcription and signal transduction. However, the biological function of sumoylation in hematopoiesis has not been fully explored. We show here that definitive hematopoietic stem/progenitor cells (HSPCs) are depleted in SUMO-deficient zebrafish embryos. Impairment of sumoylation attenuates HSPC generation and proliferation. The hyposumoylation triggered HSPC defects are CCAAT/enhancer-binding protein α (C/ebpα) dependent. Critically, a SUMO-C/ebpα fusion rescues the defective hematopoiesis in SUMO-deficient embryos, at least in part through restored runx1 expression. While C/ebpα-dependent transcription is involved in myeloid differentiation, our studies here reveal that C/ebpα sumoylation is essential for HSPC development during definitive hematopoiesis.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Diferenciação Celular/genética , Embrião não Mamífero , Técnicas de Silenciamento de Genes , Hematopoese/genética , Proteína SUMO-1/deficiência , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Sumoilação , Peixe-Zebra
6.
J Biol Chem ; 285(36): 28298-308, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20584900

RESUMO

The zinc finger transcription factor, Krüppel-like factor 4 (KLF4), regulates numerous biological processes, including proliferation, differentiation, and embryonic stem cell self-renewal. Although the DNA sequence to which KLF4 binds is established, the mechanism by which KLF4 controls transcription is not well defined. Small ubiquitin-related modifier (SUMO) is an important regulator of transcription. Here we show that KLF4 is both SUMOylated at a single lysine residue and physically interacts with SUMO-1 in a region that matches an acidic and hydrophobic residue-rich SUMO-interacting motif (SIM) consensus. The SIM in KLF4 is required for transactivation of target promoters in a SUMO-1-dependent manner. Mutation of either the acidic or hydrophobic residues in the SIM significantly impairs the ability of KLF4 to interact with SUMO-1, activate transcription, and inhibit cell proliferation. Our study provides direct evidence that SIM in KLF4 functions as a transcriptional activation domain. A survey of transcription factor sequences reveals that established transactivation domains of many transcription factors contain sequences highly related to SIM. These results, therefore, illustrate a novel mechanism by which SUMO interaction modulates the activity of transcription factors.


Assuntos
Fatores de Transcrição Kruppel-Like/química , Fatores de Transcrição Kruppel-Like/metabolismo , Proteína SUMO-1/metabolismo , Ativação Transcricional , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Bovinos , Linhagem Celular , Proliferação de Células , Chlorocebus aethiops , Técnicas de Silenciamento de Genes , Humanos , Fator 4 Semelhante a Kruppel , Lisina , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , Ratos , Proteína SUMO-1/deficiência , Proteína SUMO-1/genética
7.
J Biol Chem ; 283(46): 32045-55, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18799455

RESUMO

Small ubiquitin-like modifier (SUMO) proteases regulate the abundance and lifetime of SUMO-conjugated substrates by antagonizing reactions catalyzed by SUMO-conjugating enzymes. Six SUMO proteases constitute the human SENP/ULP protease family (SENP1-3 and SENP5-7). SENP6 and SENP7 include the most divergent class of SUMO proteases, which also includes the yeast enzyme ULP2. We present the crystal structure of the SENP7 catalytic domain at a resolution of 2.4 angstroms. Comparison with structures of human SENP1 and SENP2 reveals unique elements that differ from previously characterized structures of SUMO-deconjugating enzymes. Biochemical assays show that SENP6 and SENP7 prefer SUMO2 or SUMO3 in deconjugation reactions with rates comparable with those catalyzed by SENP2, particularly during cleavage of di-SUMO2, di-SUMO3, and poly-SUMO chains composed of SUMO2 or SUMO3. In contrast, SENP6 and SENP7 exhibit lower rates for processing pre-SUMO1, pre-SUMO2, or pre-SUMO3 in comparison with SENP2. Structure-guided mutational analysis reveals elements unique to the SENP6 and SENP7 subclass of SENP/ULP proteases that contribute to protease function during deconjugation of poly-SUMO chains.


Assuntos
Domínio Catalítico , Cisteína Endopeptidases/metabolismo , Endopeptidases/metabolismo , Proteína SUMO-1/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Cisteína Endopeptidases/genética , Endopeptidases/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , Proteína SUMO-1/deficiência , Proteína SUMO-1/genética , Alinhamento de Sequência
8.
Neuroreport ; 13(17): 2359-64, 2002 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-12488827

RESUMO

To investigate the implication of small ubiquitin-related modifier-1 (SUMO-1) in the formation of neuronal intranuclear inclusions in polyglutamine diseases, we examined the localization of SUMO-1 in dentatorubral-pallidoluysian atrophy (DRPLA) brain tissues and PC12 cells expressing truncated atrophin-1 with expanded poly-glutamine stretches. SUMO-1 was co-localized with neuronal intranuclear inclusions in DRPLA brain and the DRPLA model cells, which showed that the aggregates formed by expanded polyglutamine stretches were highly SUMOlylated. In addition, to examine the role of SUMO-1 in nuclear aggregate formation and cell death, either SUMO-1 or DeltaSUMO-1, which is a SUMOlylation defective mutant lacking the C-terminal motif, was co-transfected with atrophin-1 with expanded polyglutamine stretches. Co-transfection of DeltaSUMO-1 decreased number of the cells with nuclear aggregates and consequent apoptosis of PC12 cells, both of which were markedly enhanced by co-transfection of SUMO-1 with atrophin-1 with expanded polyglutamine stretches. These results suggest that SUMO-1 is implicated in the pathogenesis of DRPLA and accelerates aggregate formation and cell death.


Assuntos
Encéfalo/metabolismo , Morte Celular/fisiologia , Núcleo Celular/metabolismo , Corpos de Inclusão/metabolismo , Epilepsias Mioclônicas Progressivas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteína SUMO-1/deficiência , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Núcleo Celular/patologia , Humanos , Corpos de Inclusão/patologia , Pessoa de Meia-Idade , Epilepsias Mioclônicas Progressivas/genética , Epilepsias Mioclônicas Progressivas/fisiopatologia , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Células PC12 , Peptídeos/genética , Peptídeos/metabolismo , Ratos , Proteína SUMO-1/genética , Expansão das Repetições de Trinucleotídeos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...